Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 953, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38296961

RESUMEN

Autophagy is primarily activated by cellular stress, such as starvation or mitochondrial damage. However, stress-independent autophagy is activated by unclear mechanisms in several cell types, such as thymic epithelial cells (TECs). Here we report that the mitochondrial protein, C15ORF48, is a critical inducer of stress-independent autophagy. Mechanistically, C15ORF48 reduces the mitochondrial membrane potential and lowers intracellular ATP levels, thereby activating AMP-activated protein kinase and its downstream Unc-51-like kinase 1. Interestingly, C15ORF48-dependent induction of autophagy upregulates intracellular glutathione levels, promoting cell survival by reducing oxidative stress. Mice deficient in C15orf48 show a reduction in stress-independent autophagy in TECs, but not in typical starvation-induced autophagy in skeletal muscles. Moreover, C15orf48-/- mice develop autoimmunity, which is consistent with the fact that the stress-independent autophagy in TECs is crucial for the thymic self-tolerance. These results suggest that C15ORF48 induces stress-independent autophagy, thereby regulating oxidative stress and self-tolerance.


Asunto(s)
Autoinmunidad , Proteínas Mitocondriales , Ratones , Animales , Proteínas Mitocondriales/metabolismo , Estrés Oxidativo , Autofagia , Células Epiteliales/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo
2.
Genes Cells ; 28(12): 929-941, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37909727

RESUMEN

One hallmark of some autoimmune diseases is the variability of symptoms among individuals. Organs affected by the disease differ between patients, posing a challenge in diagnosing the affected organs. Although numerous studies have investigated the correlation between T cell antigen receptor (TCR) repertoires and the development of infectious and immune diseases, the correlation between TCR repertoires and variations in disease symptoms among individuals remains unclear. This study aimed to investigate the correlation of TCRα and ß repertoires in blood T cells with the extent of autoimmune signs that varies among individuals. We sequenced TCRα and ß of CD4+ CD44high CD62Llow T cells in the blood and stomachs of mice deficient in autoimmune regulator (Aire) (AIRE KO), a mouse model of human autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. Data analysis revealed that the degree of similarity in TCR sequences between the blood and stomach varied among individual AIRE KO mice and reflected the extent of T cell infiltration in the stomach. We identified a set of TCR sequences whose frequencies in blood might correlate with extent of the stomach manifestations. Our results propose a potential of using TCR repertoires not only for diagnosing disease development but also for diagnosing affected organs in autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Poliendocrinopatías Autoinmunes , Humanos , Ratones , Animales , Linfocitos T CD4-Positivos , Receptores de Antígenos de Linfocitos T/genética
3.
Front Immunol ; 14: 1186154, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38022666

RESUMEN

The thymus has the ability to regenerate from acute injury caused by radiation, infection, and stressors. In addition to thymocytes, thymic epithelial cells in the medulla (mTECs), which are crucial for T cell self-tolerance by ectopically expressing and presenting thousands of tissue-specific antigens (TSAs), are damaged by these insults and recover thereafter. However, given recent discoveries on the high heterogeneity of mTECs, it remains to be determined whether the frequency and properties of mTEC subsets are restored during thymic recovery from radiation damage. Here we demonstrate that acute total body irradiation with a sublethal dose induces aftereffects on heterogeneity and gene expression of mTECs. Single-cell RNA-sequencing (scRNA-seq) analysis showed that irradiation reduces the frequency of mTECs expressing AIRE, which is a critical regulator of TSA expression, 15 days after irradiation. In contrast, transit-amplifying mTECs (TA-mTECs), which are progenitors of AIRE-expressing mTECs, and Ccl21a-expressing mTECs, were less affected. Interestingly, a detailed analysis of scRNA-seq data suggested that the proportion of a unique mTEC cluster expressing Ccl25 and a high level of TSAs was severely decreased by irradiation. In sum, we propose that the effects of acute irradiation disrupt the heterogeneity and properties of mTECs over an extended period, which potentially leads to an impairment of thymic T cell selection.


Asunto(s)
Factores de Transcripción , Transcriptoma , Ratones , Animales , Factores de Transcripción/metabolismo , Diferenciación Celular , Ratones Endogámicos C57BL , Células Epiteliales/metabolismo
4.
Bio Protoc ; 13(1): e4588, 2023 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-36789086

RESUMEN

Accessible chromatin regions modulate gene expression by acting as cis-regulatory elements. Understanding the epigenetic landscape by mapping accessible regions of DNA is therefore imperative to decipher mechanisms of gene regulation under specific biological contexts of interest. The assay for transposase-accessible chromatin sequencing (ATAC-seq) has been widely used to detect accessible chromatin and the recent introduction of single-cell technology has increased resolution to the single-cell level. In a recent study, we used droplet-based, single-cell ATAC-seq technology (scATAC-seq) to reveal the epigenetic profile of the transit-amplifying subset of thymic epithelial cells (TECs), which was identified previously using single-cell RNA-sequencing technology (scRNA-seq). This protocol allows the preparation of nuclei from TECs in order to perform droplet-based scATAC-seq and its integrative analysis with scRNA-seq data obtained from the same cell population. Integrative analysis has the advantage of identifying cell types in scATAC-seq data based on cell cluster annotations in scRNA-seq analysis.

5.
Elife ; 112022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35578835

RESUMEN

Medullary thymic epithelial cells (mTECs) are critical for self-tolerance induction in T cells via promiscuous expression of tissue-specific antigens (TSAs), which are controlled by the transcriptional regulator, AIRE. Whereas AIRE-expressing (Aire+) mTECs undergo constant turnover in the adult thymus, mechanisms underlying differentiation of postnatal mTECs remain to be discovered. Integrative analysis of single-cell assays for transposase-accessible chromatin (scATAC-seq) and single-cell RNA sequencing (scRNA-seq) suggested the presence of proliferating mTECs with a specific chromatin structure, which express high levels of Aire and co-stimulatory molecules, CD80 (Aire+CD80hi). Proliferating Aire+CD80hi mTECs detected using Fucci technology express a minimal number of Aire-dependent TSAs and are converted into quiescent Aire+CD80hi mTECs expressing high levels of TSAs after a transit amplification. These data provide evidence for the existence of transit-amplifying Aire+mTEC precursors during the Aire+mTEC differentiation process of the postnatal thymus.


Asunto(s)
Cromatina , Análisis de la Célula Individual , Animales , Diferenciación Celular/genética , Cromatina/metabolismo , Células Epiteliales/metabolismo , Ratones , Ratones Endogámicos C57BL , Timo , Transposasas/metabolismo
6.
Pract Lab Med ; 20: e00161, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32322645

RESUMEN

Urinary gamma-glutamyltransferase (u-γGT) concentration (U/L) and excretion (urinary creatinine-corrected u-γGT; u-γGT/u-Cre, U/g creatinine) are useful markers for kidney disease. However, there is limited information available on u-γGT and u-γGT/u-Cre distribution in the elderly Japanese population. In this study, we investigated the distribution of u-γGT and u-γGT/u-Cre in 113 Japanese women aged 40-74 years. The u-γGT was assessed from spot urine samples (collected from 09:00 to 14:00) spectrophotometrically according to the Japan Society of Clinical Chemistry reference measurement procedure using l-γ-glutamyl-3-carboxy-4-nitroanilide as the substrate. The u-Cre was measured enzymatically using creatininase, creatinase, sarcosine oxidase, and peroxidase. None of the participants was diagnosed with any kidney disease. Median u-γGT and u-γGT/u-Cre values (central 95% interval values) were 29.7 (5.3-144.0) U/L and 57.9 (32.9-122.7) U/g creatinine, respectively. The distribution of u-γGT tended to decline with age. There was a statistically significant difference in the u-γGT value between the 40-59- and 60-74-year-old groups. In contrast, there was no significant difference in the u-γGT/u-Cre between each age group. The u-Cre level also declined with age. It is suggested that the decline of u-γGT with aging would be masked by the u-Cre correction.

7.
Sci Rep ; 9(1): 19866, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31882694

RESUMEN

The environment experienced during spaceflight may impact the immune system and the thymus appears to undergo atrophy during spaceflight. However, molecular aspects of this thymic atrophy remain to be elucidated. In this study, we analysed the thymi of mice on board the international space station (ISS) for approximately 1 month. Thymic size was significantly reduced after spaceflight. Notably, exposure of mice to 1 × g using centrifugation cages in the ISS significantly mitigated the reduction in thymic size. Although spaceflight caused thymic atrophy, the global thymic structure was not largely changed. However, RNA sequencing analysis of the thymus showed significantly reduced expression of cell cycle-regulating genes in two independent spaceflight samples. These reductions were partially countered by 1 × g exposure during the space flights. Thus, our data suggest that spaceflight leads to reduced proliferation of thymic cells, thereby reducing the size of the thymus, and exposure to 1 × g might alleviate the impairment of thymus homeostasis induced by spaceflight.


Asunto(s)
Gravedad Alterada , Vuelo Espacial , Timo/metabolismo , Animales , Secuencia de Bases , Ensayo de Inmunoadsorción Enzimática , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Componente Principal , RNA-Seq
8.
Commun Biol ; 2: 292, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396572

RESUMEN

Receptor activator of nuclear factor (NF)-κB (RANK) signaling promotes pregnancy-dependent epithelial cell differentiation and expansion for mammary gland development, which requires NF-κB pathway-dependent Cyclin D1 induction and inhibitor of DNA binding 2 (Id2) pathway-dependent anti-apoptotic gene induction. However, the roles of tumor necrosis factor receptor-associated factor 6 (TRAF6) remain unclear despite its requirement in RANK signaling. Here we show that TRAF6 is crucial for both mammary stem cell maintenance and pregnancy-induced epithelial cell expansion. TRAF6 deficiency impairs phosphoinositide 3-kinase (PI3K)/AKT and canonical NF-κB pathways, whereas noncanonical NF-κB signaling remains functional. Therefore, we propose that TRAF6 promotes cell proliferation by activating PI3K/AKT signaling to induce retinoblastoma phosphorylation in concert with noncanonical NF-κB pathway-dependent Cyclin D1 induction. Furthermore, TRAF6 inhibits apoptosis by activating canonical NF-κB signaling to induce anti-apoptotic genes with the Id2 pathway. Therefore, proper orchestration of TRAF6-dependent and -independent RANK signals likely establishes mammary gland formation.


Asunto(s)
Proliferación Celular , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/metabolismo , Células Madre/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/trasplante , Animales , Apoptosis , Línea Celular , Ciclina D1/metabolismo , Femenino , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones Endogámicos BALB C , Ratones Noqueados , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/deficiencia , Factor 6 Asociado a Receptor de TNF/genética
9.
Sci Rep ; 9(1): 7654, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31114014

RESUMEN

Secondary lymphoid organs are critical for regulating acquired immune responses. The aim of this study was to characterize the impact of spaceflight on secondary lymphoid organs at the molecular level. We analysed the spleens and lymph nodes from mice flown aboard the International Space Station (ISS) in orbit for 35 days, as part of a Japan Aerospace Exploration Agency mission. During flight, half of the mice were exposed to 1 g by centrifuging in the ISS, to provide information regarding the effect of microgravity and 1 g exposure during spaceflight. Whole-transcript cDNA sequencing (RNA-Seq) analysis of the spleen suggested that erythrocyte-related genes regulated by the transcription factor GATA1 were significantly down-regulated in ISS-flown vs. ground control mice. GATA1 and Tal1 (regulators of erythropoiesis) mRNA expression was consistently reduced by approximately half. These reductions were not completely alleviated by 1 g exposure in the ISS, suggesting that the combined effect of space environments aside from microgravity could down-regulate gene expression in the spleen. Additionally, plasma immunoglobulin concentrations were slightly altered in ISS-flown mice. Overall, our data suggest that spaceflight might disturb the homeostatic gene expression of the spleen through a combination of microgravity and other environmental changes.


Asunto(s)
Factor de Transcripción GATA1/metabolismo , Vuelo Espacial , Bazo/metabolismo , Transcriptoma , Animales , Regulación hacia Abajo , Eritropoyesis , Factor de Transcripción GATA1/genética , Ratones , Proteína 1 de la Leucemia Linfocítica T Aguda/genética , Proteína 1 de la Leucemia Linfocítica T Aguda/metabolismo , Ingravidez/efectos adversos
10.
Sci Rep ; 9(1): 295, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30670743

RESUMEN

Increased global incidence of myopia necessitates establishment of therapeutic approaches against its progression. To explore agents which may control myopia, we screened 207 types of natural compounds and chemical reagents based on an activity of a myopia suppressive factor, early growth response protein 1 (Egr-1) in vitro. Among the candidates, crocetin showed the highest and dose-dependent activation of Egr-1. For in vivo analysis, experimental myopia was induced in 3-week-old C57BL/6 J mice with -30 diopter (D) lenses for 3 weeks. Animals were fed with normal or mixed chow containing 0.003% (n = 19) and 0.03% (n = 7) of crocetin during myopia induction. Refraction and axial length were measured at 3-week-old and the 6-week-old with an infrared photorefractor and a SD-OCT system. Compared to controls (n = 14), crocetin administration showed a significant smaller change of refractive errors (-13.62 ± 8.14 vs +0.82 ± 5.81 D for 0.003%, p < 0.01, -2.00 ± 4.52 D for 0.03%, p < 0.01) and axial elongation (0.27 ± 0.03 vs 0.22 ± 0.04 mm for 0.003%, p < 0.01, 0.23 ± 0.05 mm for 0.03%, p < 0.05). These results suggest that a dietary factor crocetin may have a preventive effect against myopia progression.


Asunto(s)
Carotenoides/uso terapéutico , Miopía/prevención & control , Administración Oral , Animales , Carotenoides/farmacología , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Ratones , Ratones Endogámicos C57BL , Miopía/tratamiento farmacológico , Miopía/etiología , Refracción Ocular/efectos de los fármacos , Errores de Refracción/tratamiento farmacológico , Vitamina A/análogos & derivados
11.
Biochem Biophys Res Commun ; 501(3): 745-750, 2018 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-29753741

RESUMEN

Hindlimb unloading (HU) of rodents has been used as a ground-based model of spaceflight. In this study, we investigated the detailed impact of 14-day HU on the murine thymus. Thymic mass and cell number were significantly reduced after 14 days of hindlimb unloading, which was accompanied by an increment of plasma corticosterone. Although corticosterone reportedly causes selective apoptosis of CD4+CD8+ thymocytes (CD4+CD8+DPs) in mice treated with short-term HU, the reduction of thymocyte cellularity after the 14-day HU was not selective for CD4+CD8+DPs. In addition to the thymocyte reduction, the cellularity of thymic epithelial cells (TECs) was also reduced by the 14-day HU. Flow cytometric and RNA-sequencing analysis suggested that medullary TECs (mTECs) were preferentially reduced after HU. Moreover, immunohistochemical staining suggested that the 14-day HU caused a reduction of the mTECs expressing autoimmune regulator (Aire). Our data suggested that HU impacts both thymocytes and TECs. Consequently, these data imply that thymic T cell repertoire formation could be disturbed during spaceflight-like stress.


Asunto(s)
Células Epiteliales/citología , Suspensión Trasera/métodos , Timocitos/citología , Timo/fisiología , Factores de Transcripción/análisis , Animales , Antígenos CD4/análisis , Antígenos CD8/análisis , Recuento de Células , Masculino , Ratones Endogámicos C57BL , Tamaño de los Órganos , Timo/citología , Factores de Tiempo , Proteína AIRE
12.
Sci Rep ; 8(1): 2026, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29391484

RESUMEN

Despite the global pandemic of myopia, the precise molecular mechanism of the onset of myopia remains largely unknown. This is partially because of the lack of efficient murine myopic models that allow genetic manipulation at low cost. Here we report a highly practical and reproducible lens-induced myopia model by specially designed frames and lenses for mice. A lens power dependent myopic induction in mice was shown until minus 30 diopter lenses. The phenotype was significantly stronger than form-deprivation myopia. We presented the protocol for precise evaluations of the state of myopia, including refraction, corneal curvature and axial length using up-to-date devices. We also found that myopic mouse eyes showed decreased visual acuity on optokinetic response examination. Finally, we confirmed the anti-myopic effect of 1% atropine using this model, which showed its potential in drug screening. The strong phenotype, stable evaluation and the potential for gene manipulation utilizing the presented method in mice will accelerate the translational research of myopia.


Asunto(s)
Modelos Animales de Enfermedad , Miopía/patología , Animales , Lentes/efectos adversos , Ratones , Ratones Endogámicos C57BL , Miopía/etiología , Fenotipo
13.
EBioMedicine ; 15: 210-219, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28063778

RESUMEN

Prevalence of myopia is increasing worldwide. Outdoor activity is one of the most important environmental factors for myopia control. Here we show that violet light (VL, 360-400nm wavelength) suppresses myopia progression. First, we confirmed that VL suppressed the axial length (AL) elongation in the chick myopia model. Expression microarray analyses revealed that myopia suppressive gene EGR1 was upregulated by VL exposure. VL exposure induced significantly higher upregulation of EGR1 in chick chorioretinal tissues than blue light under the same conditions. Next, we conducted clinical research retrospectively to compare the AL elongation among myopic children who wore eyeglasses (VL blocked) and two types of contact lenses (partially VL blocked and VL transmitting). The data showed the VL transmitting contact lenses suppressed myopia progression most. These results suggest that VL is one of the important outdoor environmental factors for myopia control. Since VL is apt to be excluded from our modern society due to the excessive UV protection, VL exposure can be a preventive strategy against myopia progression.


Asunto(s)
Luz , Miopía/diagnóstico , Miopía/terapia , Fototerapia , Adolescente , Animales , Línea Celular , Pollos , Niño , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Anteojos , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Masculino , Miopía/etiología , Refracción Ocular , Luz Solar , Resultado del Tratamiento , Rayos Ultravioleta
14.
J Exp Med ; 213(8): 1441-58, 2016 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-27401343

RESUMEN

Medullary thymic epithelial cells (mTECs) expressing autoimmune regulator (Aire) are critical for preventing the onset of autoimmunity. However, the differentiation program of Aire-expressing mTECs (Aire(+) mTECs) is unclear. Here, we describe novel embryonic precursors of Aire(+) mTECs. We found the candidate precursors of Aire(+) mTECs (pMECs) by monitoring the expression of receptor activator of nuclear factor-κB (RANK), which is required for Aire(+) mTEC differentiation. pMECs unexpectedly expressed cortical TEC molecules in addition to the mTEC markers UEA-1 ligand and RANK and differentiated into mTECs in reaggregation thymic organ culture. Introduction of pMECs in the embryonic thymus permitted long-term maintenance of Aire(+) mTECs and efficiently suppressed the onset of autoimmunity induced by Aire(+) mTEC deficiency. Mechanistically, pMECs differentiated into Aire(+) mTECs by tumor necrosis factor receptor-associated factor 6-dependent RANK signaling. Moreover, nonclassical nuclear factor-κB activation triggered by RANK and lymphotoxin-ß receptor signaling promoted pMEC induction from progenitors exhibiting lower RANK expression and higher CD24 expression. Thus, our findings identified two novel stages in the differentiation program of Aire(+) mTECs.


Asunto(s)
Diferenciación Celular/inmunología , Células Epiteliales/inmunología , Regulación de la Expresión Génica/inmunología , Células Madre Embrionarias de Ratones/inmunología , Timo/inmunología , Factores de Transcripción/inmunología , Animales , Diferenciación Celular/genética , Células Epiteliales/citología , Regulación de la Expresión Génica/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células Madre Embrionarias de Ratones/citología , Lectinas de Plantas/genética , Lectinas de Plantas/inmunología , Timo/citología , Factores de Transcripción/genética , Proteína AIRE
15.
PLoS One ; 10(10): e0141650, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26513242

RESUMEN

Gravity change affects many immunological systems. We investigated the effects of hypergravity (2G) on murine thymic cells. Exposure of mice to 2G for three days reduced the frequency of CD4+CD8+ thymocytes (DP) and mature medullary thymic epithelial cells (mTECs), accompanied by an increment of keratin-5 and keratin-8 double-positive (K5+K8+) TECs that reportedly contain TEC progenitors. Whereas the reduction of DP was recovered by a 14-day exposure to 2G, the reduction of mature mTECs and the increment of K5+K8+ TEC persisted. Interestingly, a surgical lesion of the inner ear's vestibular apparatus inhibited these hypergravity effects. Quantitative PCR analysis revealed that the gene expression of Aire and RANK that are critical for mTEC function and development were up-regulated by the 3-day exposure and subsequently down-regulated by the 14-day exposure to 2G. Unexpectedly, this dynamic change in mTEC gene expression was independent of the vestibular apparatus. Overall, data suggest that 2G causes a temporary reduction of DP and a persistent reduction of mature mTECs in a vestibular system-dependent manner, and also dysregulates mTEC gene expression without involving the vestibular system. These data might provide insight on the impact of gravity change on thymic functions during spaceflight and living.


Asunto(s)
Recuento de Células , Células Epiteliales/metabolismo , Hipergravedad , Subgrupos de Linfocitos T/metabolismo , Timocitos/metabolismo , Timo/citología , Timo/metabolismo , Animales , Biomarcadores , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Expresión Génica , Inmunofenotipificación , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Factores de Tiempo
16.
J Biochem ; 158(6): 485-95, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26115685

RESUMEN

RelB is activated by the non-canonical NF-κB pathway, which is crucial for immunity by establishing lymphoid organogenesis and B-cell and dendritic cell (DC) maturation. To elucidate the mechanism of the RelB-mediated immune cell maturation, a precise understanding of the relationship between cell maturation and RelB expression and activation at the single-cell level is required. Therefore, we generated knock-in mice expressing a fusion protein between RelB and fluorescent protein (RelB-Venus) from the Relb locus. The Relb(Venus/Venus) mice developed without any abnormalities observed in the Relb(-/-) mice, allowing us to monitor RelB-Venus expression and nuclear localization as RelB expression and activation. Relb(Venus/Venus) DC analyses revealed that DCs consist of RelB(-), RelB(low) and RelB(high) populations. The RelB(high) population, which included mature DCs with projections, displayed RelB nuclear localization, whereas RelB in the RelB(low) population was in the cytoplasm. Although both the RelB(low) and RelB(-) populations barely showed projections, MHC II and co-stimulatory molecule expression were higher in the RelB(low) than in the RelB(-) splenic conventional DCs. Taken together, our results identify the RelB(low) population as a possible novel intermediate maturation stage of cDCs and the Relb(Venus/Venus) mice as a useful tool to analyse the dynamic regulation of the non-canonical NF-κB pathway.


Asunto(s)
Células Dendríticas/inmunología , Análisis de la Célula Individual , Factor de Transcripción ReIB/metabolismo , Animales , Linfocitos B/metabolismo , Antígeno B7-2/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Células Dendríticas/citología , Femenino , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Genes MHC Clase II , Genes Reporteros , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Bazo/citología , Timo/citología , Factor de Transcripción ReIB/genética
17.
Sci Rep ; 5: 10758, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26029823

RESUMEN

Nuclear factor (NF)-κB-inducing kinase (NIK) is a serine/threonine kinase that activates NF-κB pathways, thereby regulating a wide variety of immune systems. Aberrant NIK activation causes tumor malignancy, suggesting a requirement for precise regulation of NIK activity. To explore novel interacting proteins of NIK, we performed in vitro virus screening and identified the catalytic subunit Aα isoform of serine/threonine phosphatase calcineurin (CnAα) as a novel NIK-interacting protein. The interaction of NIK with CnAα in living cells was confirmed by co-immunoprecipitation. Calcineurin catalytic subunit Aß isoform (CnAß) also bound to NIK. Experiments using domain deletion mutants suggested that CnAα and CnAß interact with both the kinase domain and C-terminal region of NIK. Moreover, the phosphatase domain of CnAα is responsible for the interaction with NIK. Intriguingly, we found that TRAF3, a critical regulator of NIK activity, also binds to CnAα and CnAß. Depletion of CnAα and CnAß significantly enhanced lymphotoxin-ß receptor (LtßR)-mediated expression of the NIK-dependent gene Spi-B and activation of RelA and RelB, suggesting that CnAα and CnAß attenuate NF-κB activation mediated by LtßR-NIK signaling. Overall, these findings suggest a possible role of CnAα and CnAß in modifying NIK functions.


Asunto(s)
Calcineurina/metabolismo , Regulación de la Expresión Génica , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Dominio Catalítico , Línea Celular/metabolismo , Citocina TWEAK , Humanos , Isoenzimas , Receptor beta de Linfotoxina/metabolismo , Ratones , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-ets/genética , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIB/metabolismo , Factores de Transcripción/genética , Factores de Necrosis Tumoral/metabolismo , Quinasa de Factor Nuclear kappa B
18.
J Exp Med ; 211(12): 2425-38, 2014 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-25385757

RESUMEN

Medullary thymic epithelial cells (mTECs) expressing the autoimmune regulator AIRE and various tissue-specific antigens (TSAs) are critical for preventing the onset of autoimmunity and may attenuate tumor immunity. However, molecular mechanisms controlling mTEC development remain elusive. Here, we describe the roles of the transcription factor Spi-B in mTEC development. Spi-B is rapidly up-regulated by receptor activator of NF-κB ligand (RANKL) cytokine signaling, which triggers mTEC differentiation, and in turn up-regulates CD80, CD86, some TSAs, and the natural inhibitor of RANKL signaling, osteoprotegerin (OPG). Spi-B-mediated OPG expression limits mTEC development in neonates but not in embryos, suggesting developmental stage-specific negative feedback regulation. OPG-mediated negative regulation attenuates cellularity of thymic regulatory T cells and tumor development in vivo. Hence, these data suggest that this negative RANKL-Spi-B-OPG feedback mechanism finely tunes mTEC development and function and may optimize the trade-off between prevention of autoimmunity and induction of antitumor immunity.


Asunto(s)
Células Epiteliales/inmunología , Tolerancia Inmunológica/inmunología , Proteínas Proto-Oncogénicas c-ets/inmunología , Timo/inmunología , Animales , Animales Recién Nacidos , Antígeno B7-1/inmunología , Antígeno B7-1/metabolismo , Western Blotting , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Células Epiteliales/metabolismo , Retroalimentación Fisiológica , Femenino , Expresión Génica/inmunología , Tolerancia Inmunológica/genética , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/inmunología , Osteoprotegerina/metabolismo , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Ligando RANK/inmunología , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/inmunología , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Timo/metabolismo , Quinasa de Factor Nuclear kappa B
19.
PLoS One ; 9(5): e95992, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24788966

RESUMEN

Virus-derived double-stranded RNAs (dsRNAs) are sensed in the cytosol by retinoic acid-inducible gene (RIG)-I-like receptors (RLRs). These induce the expression of type I IFN and proinflammatory cytokines through signaling pathways mediated by the mitochondrial antiviral signaling (MAVS) protein. TNF receptor-associated factor (TRAF) family proteins are reported to facilitate the RLR-dependent expression of type I IFN by interacting with MAVS. However, the precise regulatory mechanisms remain unclear. Here, we show the role of FK506-binding protein 51 (FKBP51) in regulating the dsRNA-dependent expression of type I IFN. The binding of FKBP51 to TRAF6 was first identified by "in vitro virus" selection and was subsequently confirmed with a coimmunoprecipitation assay in HEK293T cells. The TRAF-C domain of TRAF6 is required for its interaction, although FKBP51 does not contain the consensus motif for interaction with the TRAF-C domain. Besides TRAF6, we found that FKBP51 also interacts with TRAF3. The depletion of FKBP51 reduced the expression of type I IFN induced by dsRNA transfection or Newcastle disease virus infection in murine fibroblasts. Consistent with this, the FKBP51 depletion attenuated dsRNA-mediated phosphorylations of IRF3 and JNK and nuclear translocation of RelA. Interestingly, dsRNA stimulation promoted the accumulation of FKBP51 in the mitochondria. Moreover, the overexpression of FKBP51 inhibited RLR-dependent transcriptional activation, suggesting a scaffolding function for FKBP51 in the MAVS-mediated signaling pathway. Overall, we have demonstrated that FKBP51 interacts with TRAF proteins and facilitates the expression of type I IFN induced by cytosolic dsRNA. These findings suggest a novel role for FKBP51 in the innate immune response to viral infection.


Asunto(s)
Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Interferón Tipo I/genética , Mitocondrias/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Línea Celular , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Ratones , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Transporte de Proteínas , ARN Bicatenario/metabolismo , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Proteínas de Unión a Tacrolimus/genética
20.
Biochem Biophys Res Commun ; 414(4): 773-8, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-22005462

RESUMEN

NF-κB-inducing kinase (NIK) plays critical roles in the development of lymph nodes and Peyer's patches, and microarchitecture of the thymus and spleen via NF-κB activation. Alymphoplasia (aly/aly) mice have a point mutation in the NIK gene that causes a defect in the activation of an NF-κB member RelB. Here, we developed a novel method to determine the aly mutation by genetic typing using PCR. This method facilitated the easy establishment of a congeneic aly/aly mouse line. Indeed, we generated a mouse line with aly mutation on a BALB/cA background (BALB/cA-aly/aly). BALB/cA-aly/aly mice showed significant splenomegaly with extramedullary hemopoiesis, which was not significant in aly/aly mice on a C57BL/6 background. Interestingly, the splenomegaly and extramedullary hemopoiesis caused by the aly mutation was gender-dependent. These data together with previous reports on extramedullary hemopoiesis in RelB-deficient mice suggest that NIK-RelB signaling may be involved in the suppression of extramedullary hemopoiesis in adult mice.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Hematopoyesis Extramedular/genética , Proteínas Serina-Treonina Quinasas/genética , Factor de Transcripción ReIB/metabolismo , Animales , Secuencia de Bases , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutación , Bazo/fisiología , Esplenomegalia/genética , Factor de Transcripción ReIB/genética , Quinasa de Factor Nuclear kappa B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...